Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Blood Adv ; 2(15): 1914-1922, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30093529

ABSTRACT

To identify novel regulators of erythropoiesis, we performed independent forward genetic screens using the chemical mutagen ENU in mice. Among progeny displaying microcytic red-cell phenotypes, 7 independent mouse strains harboring mutations within the transferrin receptor gene Tfrc were identified. Six of the mutants, including the previously described red blood cell 6 (RBC6) strain, displayed reduced erythroblast CD71 expression and midgestation lethality of homozygotes (E12.5-E14.5), and 1 novel strain, RBC21, displayed a variable phenotype with sustained CD71 expression and late homozygous lethality (E18.5). Standard iron studies were normal in the RBC21 mutant, but intracellular ferritin was significantly reduced. The microcytic phenotype seen in the RBC21 strain was the result of impaired binding of transferrin to the receptor. Neither RBC6 nor RBC21 responded to iron replacement therapy. These studies describe how point mutations of the transferrin receptor can cause a microcytic anemia that does not respond to iron therapy and would not be detected by routine iron studies, such as serum ferritin.


Subject(s)
Anemia , Antigens, CD/biosynthesis , Erythrocytes/metabolism , Ferritins/blood , Point Mutation , Receptors, Transferrin/biosynthesis , Receptors, Transferrin/genetics , Anemia/blood , Anemia/genetics , Anemia/pathology , Animals , Antigens, CD/genetics , Erythrocytes/pathology , Mice , Mice, Mutant Strains , Receptors, Transferrin/metabolism
2.
Br J Haematol ; 178(4): 616-628, 2017 08.
Article in English | MEDLINE | ID: mdl-28466468

ABSTRACT

In a dominant mouse ethylnitrosurea mutagenesis screen for genes regulating erythropoiesis, we identified a pedigree with a novel microcytic hypochromia caused by a V235G missense mutation in Dynamin 2 (Dnm2). Mutations in Dnm2, a GTPase, are highly disease-specific and have been implicated in four forms of human diseases: centronuclear myopathy, Charcot-Marie Tooth neuropathy and, more recently, T-cell leukaemia and Hereditary Spastic Paraplegia, but red cell abnormalities have not been reported to date. The V235G mutation lies within a crucial GTP nucleotide-binding pocket of Dnm2, and resulted in defective GTPase activity and incompatibility with life in the homozygous state. Dnm2 is an essential mediator of clathrin-mediated endocytosis, which is required for the uptake of transferrin (Tf) into red cells for incorporation of haem. Accordingly, we observed significantly reduced Tf uptake by Dnm2+/V235G cells, which led to impaired endosome formation. Despite these deficiencies, surprisingly all iron studies were unchanged, suggesting an unexplained alternative mechanism underlies microcytic anaemia in Dnm2+/V235G mice. This study provides the first in vivo evidence for the requirements of Dnm2 in normal erythropoiesis.


Subject(s)
Anemia, Hypochromic/genetics , Dynamin II/genetics , Mutation, Missense , Anemia, Hypochromic/blood , Animals , Chromosome Mapping/methods , Disease Models, Animal , Dynamin II/deficiency , Dynamin II/physiology , Endocytosis/genetics , Endocytosis/physiology , Erythrocytes/metabolism , Erythrocytes/pathology , Genotype , High-Throughput Nucleotide Sequencing/methods , Mice, Knockout , Transferrin/metabolism
4.
J Transl Med ; 11: 14, 2013 Jan 14.
Article in English | MEDLINE | ID: mdl-23316703

ABSTRACT

BACKGROUND: Pharmacologic reactivation of fetal hemoglobin expression is a promising strategy for treatment of sickle cell disease and ß-thalassemia. The objective of this study was to investigate the effect of the methyl transferase inhibitor adenosine-2',3'-dialdehyde (Adox) on induction of human fetal hemoglobin (HbF) in K562 cells and human hematopoietic progenitor cells. METHODS: Expression levels of human fetal hemoglobin were assessed by northern blot analysis and Real-time PCR. HbF and adult hemoglobin (HbA) content were analyzed using high-performance liquid chromatography (HPLC). DNA methylation levels on human gamma-globin gene promoters were determined using Bisulfite sequence analysis. Enrichment of histone marks on genes was assessed by chromosome immunoprecipitation (ChIP). RESULTS: Adox induced γ-globin gene expression in both K562 cells and in human bone marrow erythroid progenitor cells through a mechanism potentially involving inhibition of protein arginine methyltransferase 5 (PRMT5). CONCLUSIONS: The ability of methyl transferase inhibitors such as Adox to efficiently reactivate fetal hemoglobin expression suggests that these agents may provide a means of reactivating fetal globin expression as a therapeutic option for treating sickle cell disease and ß-thalassemia.


Subject(s)
Adenosine/analogs & derivatives , Fetal Hemoglobin/biosynthesis , Adenosine/pharmacology , Blotting, Northern , Chromatin Immunoprecipitation , Chromatography, High Pressure Liquid , Fetal Hemoglobin/genetics , Humans , K562 Cells , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction
5.
Blood Cells Mol Dis ; 50(2): 86-92, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23040355

ABSTRACT

Forward genetic screens have been performed in many species to identify phenotypes in specific organ systems. We have undertaken a large-scale N-ethyl-N-nitrosourea (ENU) mutagenesis screen to identify dominant mutations that perturb erythropoiesis in mice. Mutant mice that displayed an erythrocyte mean cell volume (MCV) greater than three standard deviations from the population mean were identified. Two of these lines, RBC13 and RBC14, displayed a hypochromic, microcytic anemia, accompanied by a marked reticulocytosis, splenomegaly and diminished red cell survival. Timed pregnancies from heterozygous intercrosses revealed that a quarter of the embryos displayed severe anemia and did not survive beyond embryonic day (E) 18.5, consistent with homozygous ß-thalassemia. Genetic complementation studies with a ß-thalassemia mouse line reproduced the embryonic lethality in compound heterozygotes and a genomic custom capture array and massively parallel sequencing of the ß-globin locus identified the causative mutations. The RBC13 line displayed a nonsense mutation at codon 40 in exon 2 of the ß-major gene, invoking parallels with the common ß(0)39 thalassemia mutation seen in humans. The RBC14 line exhibited a mutation at the polyadenylation signal of the ß-major gene, exactly replicating a human ß-thalassemia mutation. The RBC13 and RBC14 lines are the first ß-thalassemia mouse models that reproduce human ß-thalassemia at the genomic level, and as such highlight the power of ENU mutagenesis screens in generating mouse models of human disease.


Subject(s)
Disease Models, Animal , Mutagenesis , beta-Globins/genetics , beta-Thalassemia/genetics , Animals , Codon/genetics , Codon, Nonsense , Erythrocyte Indices , Ethylnitrosourea , Exons/genetics , Female , Fetal Death/genetics , Genes, Dominant , Genes, Lethal , Genetic Complementation Test , Genotype , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutagens , Polyadenylation/genetics , Pregnancy , Spleen/pathology , beta-Thalassemia/blood , beta-Thalassemia/embryology , beta-Thalassemia/pathology
6.
Haematologica ; 97(11): 1632-40, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22689669

ABSTRACT

BACKGROUND: Histone H3 lysine 4 (K4) methylation has been linked with transcriptional activity in mammalian cells. The WD40-repeat protein, WDR5, is an essential component of the MLL complex that induces histone H3 K4 methylation, but the role of WDR5 in human globin gene regulation has not yet been established. DESIGN AND METHODS: To study the role of WDR5 in human globin gene regulation, we performed knockdown experiments in both K562 cells and primary human bone marrow erythroid progenitor cells (BMC). The effects of WDR5 knockdown on γ-globin gene expression were determined. Biochemical approaches were also employed to investigate WDR5 interaction molecules. Chromosomal marks in the globin locus were analyzed by ChIP. RESULTS: We found that WDR5 interacted with protein arginine methyltransferase 5 (PRMT5), a known repressor of γ-globin gene expression, and was essential for generating tri-methylated H3K4 (H3K4me3) at the γ-globin promoter in K562 cells. Enforced expression of WDR5 in K562 cells reduced γ-globin gene expression, whereas knockdown of WDR5 increased γ-globin gene expression in both K562 cells and primary human bone marrow erythroid progenitor cells. Consistent with this, both histone H3 and H4 acetylation at the γ-globin promoter were increased, while histone H4R3 and H3K9 methylation were decreased, in WDR5 knockdown cells compared to controls. We found that WDR5 interacted with HDAC1 and a PHD domaincontaining protein, ING2 (inhibitor of growth), an H3K4me3 mark reader, to enhance γ-globin gene transcriptional repression. In human BMC, levels of WDR5 were highly enriched on the γ-promoter relative to levels on other globin promoters and compared to the γ-promoter in cord blood erythroid progenitors, suggesting that WDR5 is important in the developmental globin gene expression program. CONCLUSIONS: Our data are consistent with a model in which WDR5 binds the γ-globin promoter in a PRMT5-dependent manner; H3K4me3 induced at the γ-globin promoter by WDR5 may result in the recruitment of the ING2-associated HDAC1 component and consequent silencing of γ-globin gene expression.


Subject(s)
Erythroid Precursor Cells/metabolism , Fetal Globulins/biosynthesis , Gene Silencing/physiology , Histone-Lysine N-Methyltransferase/metabolism , Models, Biological , Promoter Regions, Genetic/physiology , Erythroid Precursor Cells/cytology , Female , Fetal Globulins/genetics , Gene Knockdown Techniques , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , K562 Cells , Male , Methylation , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
7.
Cancer Cell ; 20(5): 635-48, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22094257

ABSTRACT

Despite its prevalence, the molecular basis of squamous cell carcinoma (SCC) remains poorly understood. Here, we identify the developmental transcription factor Grhl3 as a potent tumor suppressor of SCC in mice, and demonstrate that targeting of Grhl3 by a miR-21-dependent proto-oncogenic network underpins SCC in humans. Deletion of Grhl3 in adult epidermis evokes loss of expression of PTEN, a direct GRHL3 target, resulting in aggressive SCC induced by activation of PI3K/AKT/mTOR signaling. Restoration of Pten expression completely abrogates SCC formation. Reduced levels of GRHL3 and PTEN are evident in human skin, and head and neck SCC, associated with increased expression of miR-21, which targets both tumor suppressors. Our data define the GRHL3-PTEN axis as a critical tumor suppressor pathway in SCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/metabolism , MicroRNAs/physiology , PTEN Phosphohydrolase/genetics , Skin Neoplasms/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/physiology , Animals , Base Sequence , Carcinoma, Squamous Cell/chemically induced , Cell Differentiation/genetics , Cell Proliferation , DNA-Binding Proteins/genetics , Gene Deletion , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Homeostasis , Keratinocytes/pathology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Sequence Data , Sequence Alignment , Skin Neoplasms/chemically induced , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
8.
Dev Cell ; 19(1): 138-47, 2010 Jul 20.
Article in English | MEDLINE | ID: mdl-20643356

ABSTRACT

The mammalian PCP pathway regulates diverse developmental processes requiring coordinated cellular movement, including neural tube closure and cochlear stereociliary orientation. Here, we show that epidermal wound repair is regulated by PCP signaling. Mice carrying mutant alleles of PCP genes Vangl2, Celsr1, PTK7, and Scrb1, and the transcription factor Grhl3, interact genetically, exhibiting failed wound healing, neural tube defects, and disordered cochlear polarity. Using phylogenetic analysis, ChIP, and gene expression in Grhl3(-)(/-) mice, we identified RhoGEF19, a homolog of a RhoA activator involved in PCP signaling in Xenopus, as a direct target of GRHL3. Knockdown of Grhl3 or RhoGEF19 in keratinocytes induced defects in actin polymerization, cellular polarity, and wound healing, and re-expression of RhoGEF19 rescued these defects in Grhl3-kd cells. These results define a role for Grhl3 in PCP signaling and broadly implicate this pathway in epidermal repair.


Subject(s)
Cell Polarity/physiology , Epidermis/injuries , Epidermis/physiology , Wound Healing/physiology , Actins/metabolism , Animals , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Epidermis/embryology , Female , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/physiology , Keratinocytes/cytology , Keratinocytes/physiology , Mice , Mice, Knockout , Mice, Mutant Strains , Models, Biological , Mutation , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Pregnancy , Rho Guanine Nucleotide Exchange Factors , Signal Transduction , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/physiology , Wound Healing/genetics
9.
Blood ; 116(9): 1585-92, 2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20495075

ABSTRACT

Defining the molecular mechanisms underpinning fetal (gamma) globin gene silencing may provide strategies for reactivation of gamma-gene expression, a major therapeutic objective in patients with beta-thalassemia and sickle cell disease (SCD). We have previously demonstrated that symmetric methylation of histone H4 Arginine 3 (H4R3me2s) by the protein arginine methyltransferase PRMT5 is required for recruitment of the DNA methyltransferase DNMT3A to the gamma-promoter, and subsequent DNA methylation and gene silencing. Here we show in an erythroid cell line, and in primary adult erythroid progenitors that PRMT5 induces additional repressive epigenetic marks at the gamma-promoter through the assembly of a multiprotein repressor complex containing the histone modifying enzymes SUV4-20h1, casein kinase 2alpha (CK2alpha), and components of the nucleosome remodeling and histone deacetylation complex. Expression of a mutant form of PRMT5 lacking methyltransferase activity or shRNA-mediated knockdown of SUV4-20h1 resulted in loss of complex binding to the gamma-promoter, reversal of both histone and DNA repressive epigenetic marks, and increased gamma-gene expression. The repressive H4K20me3 mark induced by SUV4-20h1 is enriched on the gamma-promoter in erythroid progenitors from adult bone marrow compared with cord blood, suggesting developmental specificity. These studies define coordinated epigenetic events linked to fetal globin gene silencing, and provide potential therapeutic targets for the treatment of beta-thalassemia and SCD.


Subject(s)
DNA Methylation , Gene Silencing , Globins/genetics , Promoter Regions, Genetic/genetics , Protein Methyltransferases/genetics , Repressor Proteins/genetics , Acetylation , Adult , Blotting, Western , Bone Marrow/metabolism , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Erythroid Precursor Cells/metabolism , Fetal Blood/metabolism , Globins/metabolism , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Immunoprecipitation , Protein Methyltransferases/antagonists & inhibitors , Protein Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
10.
Nat Struct Mol Biol ; 16(3): 304-311, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19234465

ABSTRACT

Mammalian gene silencing is established through methylation of histones and DNA, although the order in which these modifications occur remains contentious. Using the human beta-globin locus as a model, we demonstrate that symmetric methylation of histone H4 arginine 3 (H4R3me2s) by the protein arginine methyltransferase PRMT5 is required for subsequent DNA methylation. H4R3me2s serves as a direct binding target for the DNA methyltransferase DNMT3A, which interacts through the ADD domain containing the PHD motif. Loss of the H4R3me2s mark through short hairpin RNA-mediated knockdown of PRMT5 leads to reduced DNMT3A binding, loss of DNA methylation and gene activation. In primary erythroid progenitors from adult bone marrow, H4R3me2s marks the inactive methylated globin genes coincident with localization of PRMT5. Our findings define DNMT3A as both a reader and a writer of repressive epigenetic marks, thereby directly linking histone and DNA methylation in gene silencing.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Gene Silencing , Histones/metabolism , Protein Methyltransferases/metabolism , Arginine/metabolism , DNA Methyltransferase 3A , Erythroid Precursor Cells/chemistry , Gene Knockdown Techniques , Humans , Models, Biological , Protein Binding , Protein Interaction Domains and Motifs , Protein-Arginine N-Methyltransferases
11.
Blood ; 113(14): 3352-62, 2009 Apr 02.
Article in English | MEDLINE | ID: mdl-19179303

ABSTRACT

Insights into the role of ankyrin-1 (ANK-1) in the formation and stabilization of the red cell cytoskeleton have come from studies on the nb/nb mice, which carry hypomorphic alleles of Ank-1. Here, we revise several paradigms established in the nb/nb mice through analysis of an N-ethyl-N-nitrosourea (ENU)-induced Ank-1-null mouse. Mice homozygous for the Ank-1 mutation are profoundly anemic in utero and most die perinatally, indicating that Ank-1 plays a nonredundant role in erythroid development. The surviving pups exhibit features of severe hereditary spherocytosis (HS), with marked hemolysis, jaundice, compensatory extramedullary erythropoiesis, and tissue iron overload. Red cell membrane analysis reveals a complete loss of ANK-1 protein and a marked reduction in beta-spectrin. As a consequence, the red cells exhibit total disruption of cytoskeletal architecture and severely altered hemorheologic properties. Heterozygous mutant mice, which have wild-type levels of ANK-1 and spectrin in their RBC membranes and normal red cell survival and ultrastructure, exhibit profound resistance to malaria, which is not due to impaired parasite entry into RBC. These findings provide novel insights into the role of Ank-1, and define an ideal model for the study of HS and malarial resistance.


Subject(s)
Ankyrins/physiology , Erythroid Cells/metabolism , Ethylnitrosourea , Hematologic Neoplasms/chemically induced , Hematologic Neoplasms/genetics , Animals , Animals, Newborn , Ankyrins/genetics , Ankyrins/metabolism , Base Sequence , Carcinogens , Cytoskeleton/genetics , Cytoskeleton/pathology , DNA Mutational Analysis , Erythrocytes/drug effects , Erythrocytes/pathology , Erythrocytes, Abnormal/pathology , Erythropoiesis/genetics , Erythropoiesis/physiology , Hematologic Neoplasms/pathology , Hemolysis/drug effects , Hemolysis/genetics , Malaria/genetics , Malaria/veterinary , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data
12.
Blood ; 107(5): 2138-45, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16263792

ABSTRACT

Binding of the stage selector protein (SSP) to the stage selector element (SSE) in the human gamma-globin promoter contributes to the preferential expression of the gamma-gene in fetal erythroid cells. The SSP contains the transcription factor CP2 and an erythroid-specific partner, NF-E4. The NF-E4 gene encodes a 22-kDa polypeptide employing a non-AUG initiation codon. Antisera specific to NF-E4 detects this species and an additional 14 kDa protein, which initiates from an internal methionine. Enforced expression of p14 NF-E4 in the K562 fetal/erythroid cell line, and in primary erythroid cord blood progenitors, results in repression of gamma-gene expression. Biochemical studies reveal that p14 NF-E4 interacts with CP2, resulting in diminished association of CP2 with the SSE in chromatin immunoprecipitation assays. p45 NF-E2 recruitment to the gamma-promoter is also lost, resulting in a reduction in RNA polymerase II and TBP binding and a fall in promoter transcriptional activity. This effect is specific, as enforced expression of a mutant form of p14 NF-E4, which fails to interact with CP2, also fails to repress gamma-gene expression in K562 cells. These findings provide one potential mechanism that could contribute to the autonomous silencing of the human gamma-genes in adult erythroid cells.


Subject(s)
Fetal Blood/physiology , Gene Silencing/physiology , Globins/biosynthesis , NF-E2 Transcription Factor, p45 Subunit/genetics , Transcription Factors/genetics , Erythroid Precursor Cells , Fetal Blood/cytology , Globins/genetics , Humans , K562 Cells , NF-E2 Transcription Factor, p45 Subunit/deficiency , NF-E2 Transcription Factor, p45 Subunit/metabolism , Protein Binding/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Response Elements/genetics , Transcription Factors/metabolism , Transduction, Genetic/methods
13.
Mol Cell Biol ; 22(22): 8026-34, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12391168

ABSTRACT

The proteins of the trithorax and Polycomb groups maintain the differential expression pattern of homeotic genes established by the early embryonic patterning system during development. These proteins generate stable and heritable chromatin structures by acting via particular chromosomal memory elements. We established a transgenic assay system showing that the Polycomb group response elements bxd and Mcp confer epigenetic inheritance throughout development. With previously published data for the Fab7 cellular memory module, we confirmed the cellular memory function of Polycomb group response elements. In Drosophila melanogaster, several of these memory elements are located in the large intergenic regulatory regions of the homeotic bithorax complex. Using a transgene assay, we showed that transcription through a memory element correlated with the relief of silencing imposed by the Polycomb group proteins and established an epigenetically heritable active chromatin mode. A memory element remodeled by the process of transcription was able to maintain active expression of a reporter gene throughout development. Thus, transcription appears to reset and change epigenetic marks at chromosomal memory elements regulated by the Polycomb and trithorax proteins. Interestingly, in the bithorax complex of D. melanogaster, the segment-specific expression of noncoding intergenic transcripts during embryogenesis seems to fulfill this switching role for memory elements regulating the homeotic genes.


Subject(s)
DNA, Intergenic/genetics , Drosophila melanogaster/genetics , Genes, Homeobox/genetics , Transcription Factors , Transcription, Genetic , Animals , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/embryology , Embryo, Nonmammalian/anatomy & histology , Embryo, Nonmammalian/physiology , Genes, Insect , Genes, Reporter , In Situ Hybridization , Models, Genetic , Organisms, Genetically Modified , Polycomb Repressive Complex 1 , Regulatory Sequences, Nucleic Acid , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...